Development of N-Hydroxycinnamamide-Based Histone Deacetylase

Jan 8, 2013 - Postdoctoral Workstation, Biomedical Industry Park Management Office, Weifang, Shandong, 261205, People,s Republic of China...
0 downloads 0 Views 1MB Size
Letter pubs.acs.org/acsmedchemlett

Development of N‑Hydroxycinnamamide-Based Histone Deacetylase Inhibitors with an Indole-Containing Cap Group Yingjie Zhang,†,⊥ Penghui Yang,†,⊥ C. James Chou,‡ Chunxi Liu,§ Xuejian Wang,∥ and Wenfang Xu*,† †

Department of Medicinal Chemistry, School of Pharmacy, Shandong University, Ji’nan, Shandong, 250012, People’s Republic of China ‡ Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, Medical University of South Carolina, Charleston, South Carolina, 29425, United States United States § Department of Pharmaceutics, School of Pharmacy, Shandong University, Ji’nan, Shandong, 250012, People’s Republic of China ∥ Postdoctoral Workstation, Biomedical Industry Park Management Office, Weifang, Shandong, 261205, People’s Republic of China S Supporting Information *

ABSTRACT: A novel series of histone deacetylase inhibitors combining Nhydroxycinnamamide bioactive fragment and indole bioactive fragment was designed and synthesized. Several compounds (17c, 17g, 17h, 17j, and 17k) exhibited comparable, even superior, total HDACs inhibitory activity and in vitro antiproliferative activities relative to the approved drug SAHA. A representative compound 17a with moderate HDACs inhibition was progressed to isoform selectivity profile, Western blot analysis, and in vivo antitumor assay. Although HDACs isoform selectivity of 17a was similar to that of SAHA, our Western blot results indicated that intracellular effects of 17a at 1 μM were class I selective. It was noteworthy that the effect on histone H4 acetylation of SAHA decreased with time, while the effect on histone H4 acetylation of 17a was maintained and even increased. Most importantly, compound 17a exhibited promising in vivo antitumor activity in a U937 xenograft model. KEYWORDS: histone deacetylases, inhibitor, N-hydroxycinnamamide, indole

T

he epigenetic regulation of histone and DNA plays an important role in chromatin structure and control of gene expression. Altered patterns of epigenetic modification are very common in many diseases, including cancer.1 As with genetic information, epigenetic modifications are heritable; in contrast, however, they are reversible and catalyzed by pairs of enzymes with converse activity. Among these epigenetic enzymes, histone deacetylases (HDACs) have now emerged as a promising new class of therapeutic targets.2 Four classes were identified in the HDACs family, characterized by different cellular localization and substrate specificity. The common feature of classes I (HDAC1−3 and -8), II (HDAC4−7, -9, and -10) and IV (HDAC11) active sites are characterized by the presence of a catalytic Zn2+ ion, and the class III HDACs (sirtuins 1−7) are NAD+-dependent. These HDACs isoforms perform their multiple functions by either epigenetic mechanism (deacetylation of histones) or nonepigenetic mechanism (deacetylation of nonhistone substrates). It has been revealed that Zn2+-dependent isozymes, especially class I and class II, are closely related to tumorigenesis and development.3 In the past 10 years, over 490 clinical trials of more than 20 histone deacetylase inhibitor (HDACI) candidates have been initiated, culminating in the approval of two antitumor drugs, vorinostat (SAHA) and romidepsin (FK228) (Figure 1).4,5 © 2013 American Chemical Society

Figure 1. Approved and clinical HDACIs with the N-hydroxycinnamamide fragment highlighted in red.

N-Hydroxycinnamamide (Figure 1) is a very common active fragment in HDACI design.6,7 This fragment could not only form bichelation with the active site Zn2+ by its hydroxamic acid group but also form a sandwichlike π−π interaction by inserting its vinyl benzene group into two parallel phenylalanine residues of HDAC. Currently, there are three HDACIs Received: October 27, 2012 Accepted: January 8, 2013 Published: January 8, 2013 235

dx.doi.org/10.1021/ml300366t | ACS Med. Chem. Lett. 2013, 4, 235−238

ACS Medicinal Chemistry Letters

Letter

containing N-hydroxycinnamamide fragment in clinical trials (Figure 1). Indole is recognized as a privileged structure in drug design,8,9 and its derivatives have been found to exhibit anticancer activity by interacting with different targets.10 Kinds of natural and synthetic HDACIs containing an indole scaffold, especially in the cap group, possessed promising in vitro and in vivo potency.11−14 Recently, comprehensive HDACIs structure−activity relationship (SAR) studies revealed that Nhydroxycinnamamide-based compounds were more stable than their straight chain analogues, and compounds having indole groups exhibited the best in vivo efficacy and tolerability.15 On the basis of the aforementioned information, we designed a novel series of N-hydroxycinnamamide-based HDACIs with an indole-containing cap group. Compounds 14a, 14b, and 15 were synthesized following the procedures described in Scheme 1. Methyl ester protection and Scheme 1. Synthesis of Compounds 14a, 14b, and 15

Table 1. HeLa Cell Nuclear Extract Inhibitory Activity compd

IC50 (μM)a

compd

14a 14b 15 17a 17b 17c 17d 17e 17f 17g 17h 17i SAHA

0.65 2.09 2.91 1.08 0.49 0.17 0.62 0.57 0.38 0.16 0.18 0.43 0.19

± ± ± ± ± ± ± ± ± ± ± ± ±

17j 17k 17l 17m 17n 17o 17p 17q 17r 17s 17t 17u

0.12 0.31 0.34 0.20 0.09 0.04 0.13 0.09 0.05 0.04 0.04 0.05 0.03

IC50 (μM)a 0.18 0.14 0.32 0.43 1.17 0.80 1.12 0.73 b 1.06 1.47 0.71

± ± ± ± ± ± ± ±

0.03 0.03 0.05 0.05 0.21 0.17 0.23 0.16

± 0.20 ± 0.36 ± 0.19

Assays were performed in replicate (n ≥ 2). Data are shown as means ± SDs. bCompound 17r was undissolved under our test condition.

a

a

Scheme 2. Synthesis of Compounds 17a−ua

a Reagents and conditions: (a) SOCI2, CH3OH, 98%. (b) (Boc)2O, Et3N, DCM, 80%. (c) LiAIH4, anhydrous THF, 0 °C, 86%. (d) PTSA, CH3OH, 80 °C, 96%. (e) H2, 10% Pd−C, CH3OH. (f) PPh3, DEAD, anhydrous THF, 0 °C, 75% for 8a, 79% for 8b. (g) NH2OK, CH3OH, 51% for 9a, 49% for 9b. (h) HCI, anhydrous EtOAc, 90%.

tert-butyloxycarbonyl (Boc) protection of L-tryptophan (1) followed by LiAlH4 reduction provided the intermediate 9. Methyl ester protection of ferulic acid (10) afforded compound 11, which was hydrogenized to give compound 12. Both 11 and 12 could be connected with 9 under Mitsunobu reaction conditions to get 13a and 13b, which were converted to corresponding hydroxamic acid compounds 14a and 14b, respectively. Subsequent N-deprotection of 14a afforded compound 15. The preliminary HDACs inhibitory assay was tested against HeLa cell nuclear extract, mainly containing HDAC1 and HDAC2. Results listed in Table 1 revealed that compound 14a (IC50 = 0.65 μM) was more potent than its analogues 14b (IC50 = 2.09 μM) and 15 (IC50 = 2.91 μM), which indicated that both the vinyl benzene group and the substituent located on the amine atom (the Boc group in 14a) were advantageous to HDACs inhibition. Therefore, using 14a as lead, we kept its N-hydroxycinnamamide group unchanged and derivatized its Boc group to other functional groups. Such derivatizations were performed according to the methods in Scheme 2. NDeprotection of 13a with trifluoroacetic acid (TFA) and subsequent amide condensation or sulfonylation gave intermediates 16a−u, which were treated with NH2OK to get target compounds 17a−u. As shown in Table 1, we found that compounds 17b−n with N-Boc-protected natural α-amino acid residues as R group except 17n exhibited more potent activity than their parent compound 14a. Most importantly, the total HDACs inhibitory

a Reagents and conditions: (a) (i) TFA, Et3N, DCM; (ii) R′COOH, TBTU, Et3N, THF, 52−75% for two steps. (b) (i) TFA, Et3N, DCM; (ii) R′SO2CI, Et3N, DCM, 60−76% for two steps. (c) NH2OK, CH3OH, 31−54%.

activities of compounds 17c, 17g, 17h, 17j, and 17k were comparable, even more potent than that of SAHA. However, replacing the Boc group of 14a with valproyl group (17a), unnatural amino acid residues (17o−q) and sulfuryl group (17r−u) were detrimental to activity. To characterize HDACs isoform selectivity of these analogues, representative compound 17a with moderate total HDACs inhibitory activity was tested against HDAC1, HDAC2, HDAC3, and HDAC6 using acetylated substrate. Besides, the class IIa inhibitory activity was evaluated against MDA-MB-231 cell lysate using class IIa-specific triflouroacetylated substrate.16 Results in Table 2 showed that 17a displayed modest preference for HDAC1 and HDAC3 over HDAC2 and HDAC6 but exhibited no obvious inhibition against class IIa HDACs up to 10 μM. The overall selectivity profile of 17a was 236

dx.doi.org/10.1021/ml300366t | ACS Med. Chem. Lett. 2013, 4, 235−238

ACS Medicinal Chemistry Letters

Letter

Table 2. HDACs Isoform Selectivity of 17a and SAHAa class I

a

class IIb

class IIa

compd

HDAC1

HDAC2

HDAC3

HDAC6

cell lysate

17a SAHA

0.39 ± 0.12 0.076 ± 0.011

1.42 ± 0.06 0.256 ± 0.003

0.28 ± 0.13 0.028 ± 0.011

0.94 ± 0.14 0.118 ± 0.012

NAb NAb

Assays were performed in replicate (n ≥ 2). IC50 (μM) values are shown as means ± SDs. bNA, not active at 10 μM.

similar to that of SAHA, which was in line with literature information.17

Table 3. In Vitro Antiproliferative Activity of Representative Compounds IC50 (μM)a compd

U937

PC-3

A549

ES-2

MDA-MB-231

HCT116

17a 17c 17g 17h 17j 17k SAHA

1.8 3.1 2.2 2.2 2.7 3.9 2.3

3.7 10.5 10.4 5.8 5.4 8.2 9.9

4.4 11.8 4.2 1.6 7.0 13.5 3.8

5.4 29.2 25.1 4.4 8.9 9.5 12.7

3.1 7.2 4.5 6.8 7.2 11.7 5.6

5.5 6.0 3.8 5.9 2.4 9.4 6.0

a

Values are the means of at least two experiments. The SD values are